ABSTRACT
This study aimed to evaluate the safety profiles of FcRn antagonists, efgartigimod alfa and rozanolixizumab, in the treatment of myasthenia gravis using real-world adverse event data from the FAERS database. A disproportionality analysis was conducted employing Reporting Odds Ratio (ROR) and Proportional Reporting Ratio (PRR) methods on reports from Q1 2022 to Q2 2025. The most frequently reported adverse events for efgartigimod alfa included falls, urinary tract infections, and symptom recurrence, with signals also detected for atrial fibrillation, peripheral neuropathy, and prostate cancer. For rozanolixizumab, common events were headache, diarrhea, and vomiting, with potential signals such as meningitis, hypersomnia, and feeding disorder. Subgroup analysis revealed gender-specific differences in adverse reactions. Most events occurred within 30 days of treatment initiation, though efgartigimod alfa showed a sustained risk beyond 180 days. The study identifies novel safety signals and highlights clinically relevant risk patterns, supporting enhanced monitoring in clinical use. It is crucial to emphasize that this disproportionality analysis is exploratory in nature and identifies potential associations, which do not establish causality and require confirmation through further dedicated studies.
KEYWORDS: Efgartigimod alfa, rozanolixizumab, FcRn antagonists, disproportionality analysis, FAERS
Introduction
Generalized myasthenia gravis (gMG) is a chronic autoimmune disorder characterized by fatigable weakness of skeletal muscles, primarily mediated by pathogenic immunoglobulin G (IgG) autoantibodies targeting the neuromuscular junction.1–3 The therapeutic landscape for gMG has been significantly transformed by the advent of targeted biologic agents designed to specifically interrupt autoimmune pathways.4 Among these innovations, neonatal Fc receptor (FcRn) antagonists constitute a novel therapeutic class with a distinct mechanism of action.5 By inhibiting the FcRn-IgG interaction, these compounds accelerate the catabolism of pathogenic IgG antibodies, reducing their serum concentrations and alleviating the autoimmune assault on neuromuscular transmission.6–8 Efgartigimod alfa and rozanolixizumab are pioneering FcRn antagonists that have gained regulatory approval or are under extensive clinical evaluation for gMG, presenting a promising therapeutic option for patients with refractory disease.9,10
Although clinical trials have established the efficacy of FcRn antagonists in reducing disease severity and improving patient outcomes,11,12 the comprehensive safety profile of these biologics in real-world clinical practice remains incompletely elucidated. Pre-marketing studies are inherently constrained by their finite duration, selective patient cohorts, and controlled environments, potentially overlooking rare, delayed, or demographic-specific adverse events (AEs). Consequently, robust post-marketing surveillance is imperative for identifying unforeseen safety signals. The US FDA Adverse Event Reporting System (FAERS) database is a vital resource for such pharmacovigilance endeavors, aggregating spontaneous AE reports that facilitate the detection of potential drug-AE associations through established statistical methodologies like disproportionality analysis.13
Focusing on the FcRn antagonists efgartigimod alfa and rozanolixizumab, initial clinical trials indicated generally tolerable safety profiles, with commonly reported AEs including headache, infections, and administration-site reactions.14–16 Nonetheless, the long-term and broader spectrum of AEs, especially serious and unanticipated ones, requires continued evaluation in heterogeneous, real-world populations. Moreover, potential differences in safety signals between these two agents – possibly attributable to variations in their molecular structures, dosing protocols, or detailed mechanisms – are not yet well-defined. Disproportionality analysis, employing measures such as the Reporting Odds Ratio (ROR) and Proportional Reporting Ratio (PRR), provides a powerful approach for mining the FAERS database to uncover such signals, highlighting AEs reported with disproportionately high frequency for a target drug compared to all other agents in the database.
Despite their growing incorporation into clinical practice, a comprehensive, direct comparative safety assessment of efgartigimod alfa and rozanolixizumab utilizing large-scale real-world data is absent.10 Critical questions remain unanswered: What are the most prevalent AEs reported for each drug in a real-world setting? Are there novel, unexpected safety signals not previously captured in controlled trials? Do the AE profiles differ meaningfully between the two therapeutics? Do specific patient subgroups, defined by factors such as gender, exhibit heightened vulnerability to particular AEs? Additionally, elucidating the temporal pattern of AE onset following treatment initiation is essential for formulating effective clinical monitoring guidelines. Addressing these questions is paramount for refining risk-benefit evaluations and tailoring patient management strategies for these novel biologics.
This study therefore utilized the FAERS database to conduct a detailed disproportionality analysis of adverse events associated with efgartigimod alfa and rozanolixizumab following their introduction. We aimed to characterize and compare their real-world safety profiles, identify and validate potential new safety signals, investigate gender-based disparities in adverse reactions, and analyze the time-to-onset patterns of reported AEs. Our analysis provides a essential post-marketing safety evaluation, uncovering distinct adverse event patterns, revealing novel potential risks such as atrial fibrillation and peripheral neuropathy for efgartigimod, and meningitis and hypersomnia for rozanolixizumab, and highlighting subgroup susceptibilities and temporal trends. These findings significantly advance the understanding of the safety landscapes of these FcRn antagonists, delivering valuable insights to inform their clinical use and guide future safety investigations.
Materials and methods
Data source and processing
This study retrieved adverse event (AE) data from the US FDA Adverse Event Reporting System (FAERS) database covering the period from the first quarter of 2013 to the second quarter of 2025. Data processing adhered to the standard methodology recommended by the Food and Drug Administration (FDA): the PRIMARYID, CASEID, and FDA_DT fields were initially extracted from the DEMO table and sorted sequentially by CASEID, FDA_DT, and PRIMARYID. For reports sharing identical CASEID values, only the record with the latest FDA_DT value was retained. In cases where both CASEID and FDA_DT were identical, the entry with the highest PRIMARYID value was selected. All adverse events were coded using the MedDRA version 28.0 terminology set for standardization.17
Data analysis
In this study, we employed multiple pharmacovigilance methodologies for analysis, including the Reporting Odds Ratio (ROR) and Proportional Reporting Ratio (PRR).18,19 Our investigation required the simultaneous satisfaction of both algorithms’ positive thresholds for an adverse event to be identified as a potential signal. Supplementary Table S1 provides a detailed 2 × 2 contingency matrix, while Supplementary Table S2 specifies the concrete parameters of the two primary signal detection algorithms.
Results
Descriptive analysis
We analyzed adverse event (AE) reports associated with efgartigimod alfa (n = 2,519) and rozanolixizumab (n = 442). The baseline clinical characteristics of the reported patients are summarized in Table 1. Female patients accounted for a higher proportion of reports than males for both therapeutics, and the most frequently reported body weight range was 50–100 kg. For efgartigimod alfa, the most commonly reported age group was greater than 65 years, whereas for rozanolixizumab, it was 18–65 years. The United States contributed the highest number of reports for both drugs, and consumer reports constituted the largest proportion. Since 2023, the number of reports for both agents has increased annually.
Table 1.
Demographic information reported by AE in FAERS.
| Efgartigimod alfa (n = 2519) |
Rozanolixizumab (n = 442) |
|||||
|---|---|---|---|---|---|---|
| n | % | n | % | |||
| Sex | Female | 211 | 8.4% | Female | 251 | 56.8% |
| Male | 188 | 7.5% | Male | 175 | 39.6% | |
| Missing | 2120 | 84.1% | Missing | 16 | 3.6% | |
| Weight(kg) | <50 | 12 | 0.5% | <50 | 11 | 2.5% |
| >100 | 54 | 2.1% | >100 | 23 | 5.2% | |
| 50~100 | 127 | 5.1% | 50~100 | 72 | 16.3% | |
| Missing | 2326 | 92.3% | Missing | 336 | 76.0% | |
| Age(years) | <18 | 3 | 0.1% | <18 | 5 | 1.1% |
| 18~64.9 | 95 | 3.8% | 18~64.9 | 110 | 24.9% | |
| >65 | 129 | 5.1% | >65 | 87 | 19.7% | |
| Missing | 2292 | 91.0% | Missing | 240 | 54.3% | |
| Outcome | Death | 227 | 9.0% | Death | 8 | 1.8% |
| Disability | 5 | 0.2% | Disability | 1 | 0.2% | |
| Hospitalization | 1077 | 42.8% | Hospitalization | 139 | 31.4% | |
| Life-Threatening | 164 | 6.5% | Life-Threatening | 6 | 1.4% | |
| Other | 1046 | 41.5% | Other | 288 | 65.2% | |
| Reporter | Consumer | 1840 | 73.0% | Consumer | 258 | 58.4% |
| Health Professional | 266 | 10.6% | Health Professional | 104 | 23.5% | |
| Pharmacist | 67 | 2.7% | Pharmacist | 23 | 5.2% | |
| Physician | 332 | 13.2% | Physician | 57 | 12.9% | |
| Missing | 14 | 0.6% | ||||
| Top 5 Reported Countries | United States | 2045 | 81.2% | United States | 381 | 86.2% |
| Japan | 243 | 9.6% | Japan | 37 | 8.4% | |
| Germany | 40 | 1.6% | Germany | 11 | 2.5% | |
| China | 33 | 1.3% | Italy | 6 | 1.3% | |
| Great Britain | 28 | 1.1% | France | 5 | 1.1% | |
| Other | 130 | 5.2% | Australia | 2 | 0.5% | |
| Reporting years | 2022 | 22 | 0.9% | 2023 | 20 | 4.5% |
| 2023 | 957 | 38.0% | 2024 | 210 | 47.5% | |
| 2024 | 1337 | 53.1% | 2025 | 212 | 48.0% | |
| 2025 | 203 | 8.1% | ||||
Signal detection based on SOC levels
Table 2 presents the signal strengths of adverse reactions at the System Organ Class (SOC) level for both efgartigimod alfa and rozanolixizumab. For efgartigimod alfa, the only category meeting both algorithmic criteria (ROR and PRR) was Surgical and medical procedures (ROR = 4.53, 95% CI: 4.06–5.06; PRR = 4.35, x2 = 867.58), while the three SOCs with the highest incidence of adverse reactions were General disorders and administration site conditions (n = 2,236), Nervous system disorders (n = 1,834), and Infections and infestations (n = 1,287). Similarly, for rozanolixizumab, the only SOC identified by both algorithms was also Surgical and medical procedures (ROR = 2.14, 95% CI: 1.69–2.69; PRR = 2.09, x2 = 43.07), with its most frequently reported SOCs being General disorders and administration site conditions (n = 448), Nervous system disorders (n = 392), and Gastrointestinal disorders (n = 203). These results indicate specific safety profiles that merit clinical attention for each agent.
Table 2.
Distribution of signal strength for adverse event reports of FcRn antagonists at the System Organ Class (SOC) level in the FAERS database.
| SOC | efgartigimod alfa |
Rozanolixizumab |
||||
|---|---|---|---|---|---|---|
| n | ROR(95%Cl) | PRR(x2) | n | ROR(95%Cl) | PRR(x2) | |
| BLOOD AND LYMPHATIC SYSTEM DISORDERS | 54 | 0.33(0.25–0.44) | 0.34(66.61) | 6 | 0.25(0.11–0.56) | 0.25(13.17) |
| CARDIAC DISORDERS | 186 | 0.91(0.77–1.06) | 0.91(1.52) | 27 | 0.81(0.55–1.19) | 0.82(1.12) |
| CONGENITAL, FAMILIAL AND GENETIC DISORDERS | 4 | 0.43(0.15–1.2) | 0.43(2.74) | |||
| EAR AND LABYRINTH DISORDERS | 40 | 0.98(0.7–1.39) | 0.98(0.01) | 7 | 1.06(0.5–2.25) | 1.06(0.02) |
| ENDOCRINE DISORDERS | 29 | 1.09(0.72–1.63) | 1.09(0.16) | |||
| EYE DISORDERS | 505 | 1.04(0.94–1.14) | 1.03(0.47) | 65 | 0.8(0.62–1.03) | 0.81(3.11) |
| GASTROINTESTINAL DISORDERS | 732 | 0.86(0.8–0.94) | 0.87(12.47) | 203 | 1.6(1.38–1.86) | 1.54(39.34) |
| GENERAL DISORDERS AND ADMINISTRATION SITE CONDITIONS | 2236 | 0.87(0.83–0.92) | 0.9(28) | 448 | 1.16(1.04–1.29) | 1.13(7.54) |
| HEPATOBILIARY DISORDERS | 46 | 0.79(0.58–1.08) | 0.79(2.15) | 1 | 0.11(0.01–0.76) | 0.11(7.49) |
| IMMUNE SYSTEM DISORDERS | 79 | 0.68(0.53–0.86) | 0.68(10.54) | 11 | 0.61(0.33–1.1) | 0.61(2.76) |
| INFECTIONS AND INFESTATIONS | 1287 | 1.27(1.19–1.36) | 1.25(53.67) | 140 | 0.77(0.65–0.92) | 0.79(8.4) |
| INJURY, POISONING AND PROCEDURAL COMPLICATIONS | 948 | 0.83(0.77–0.89) | 0.84(24.96) | 125 | 0.68(0.56–0.81) | 0.7(17.89) |
| INVESTIGATIONS | 467 | 0.68(0.62–0.75) | 0.7(56.91) | 59 | 0.55(0.42–0.71) | 0.56(20.86) |
| METABOLISM AND NUTRITION DISORDERS | 145 | 0.77(0.64–0.91) | 0.77(8.83) | 16 | 0.53(0.32–0.87) | 0.53(6.49) |
| MUSCULOSKELETAL AND CONNECTIVE TISSUE DISORDERS | 783 | 0.83(0.77–0.9) | 0.84(20.81) | 112 | 0.74(0.61–0.9) | 0.75(9.43) |
| NEOPLASMS BENIGN, MALIGNANT AND UNSPECIFIED (INCL CYSTS AND POLYPS) | 185 | 1.27(1.08–1.5) | 1.27(8.25) | 10 | 0.39(0.21–0.73) | 0.39(9.38) |
| NERVOUS SYSTEM DISORDERS | 1834 | 1.28(1.21–1.35) | 1.23(73.12) | 392 | 1.72(1.53–1.92) | 1.58(90.21) |
| PREGNANCY, PUERPERIUM AND PERINATAL CONDITIONS | 2 | 0.1(0.02–0.39) | 0.1(16.62) | |||
| PRODUCT ISSUES | 27 | 0.32(0.21–0.47) | 0.32(37.04) | 20 | 1.69(1.07–2.65) | 1.68(5.28) |
| PSYCHIATRIC DISORDERS | 262 | 0.84(0.74–0.96) | 0.84(6.55) | 34 | 0.68(0.48–0.96) | 0.68(5) |
| RENAL AND URINARY DISORDERS | 175 | 1.23(1.04–1.46) | 1.23(5.92) | 12 | 0.48(0.27–0.86) | 0.49(6.43) |
| REPRODUCTIVE SYSTEM AND BREAST DISORDERS | 33 | 1.28(0.87–1.88) | 1.28(1.55) | 3 | 0.67(0.21–2.1) | 0.67(0.48) |
| RESPIRATORY, THORACIC AND MEDIASTINAL DISORDERS | 920 | 1.29(1.2–1.39) | 1.27(43.77) | 110 | 0.88(0.72–1.06) | 0.88(1.76) |
| SKIN AND SUBCUTANEOUS TISSUE DISORDERS | 252 | 0.57(0.5–0.65) | 0.58(70.22) | 65 | 0.99(0.77–1.27) | 0.99(0) |
| SOCIAL CIRCUMSTANCES | 110 | 1.84(1.47–2.3) | 1.83(29.72) | 20 | 1.82(1.16–2.87) | 1.82(6.98) |
| SURGICAL AND MEDICAL PROCEDURES | 647 | 4.53(4.06–5.06) | 4.35(867.58) | 79 | 2.14(1.69–2.69) | 2.09(43.07) |
| VASCULAR DISORDERS | 182 | 0.9(0.77–1.06) | 0.91(1.52) | 22 | 0.67(0.44–1.03) | 0.68(3.41) |
Signal detection based on PT levels
As shown in Table 3, in the analysis at the Preferred Term (PT) level, we first screened for PTs that met the criteria of both algorithms (ROR and PRR), subsequently excluding terms unrelated to adverse drug reactions, ultimately identifying statistically significant signals. For efgartigimod alfa, common adverse reactions included falls (n = 173), urinary tract infection (n = 160), and symptom recurrence (n = 157). Furthermore, several potential adverse reactions with signal strength were identified, including atrial fibrillation (ROR = 2.15, 95% CI: 1.50–3.07; PRR = 2.14, x2 = 18.23), peripheral neuropathy (ROR = 2.21, 95% CI: 1.31–3.73; PRR = 2.21, x2 = 9.37), and prostate cancer (ROR = 2.56, 95% CI: 1.30–5.06; PRR = 2.56, x2 = 7.89). For rozanolixizumab, common adverse reactions included headache (n = 86), diarrhea (n = 47), and vomiting (n = 31). Concurrently, potential signals were detected, such as meningitis (ROR = 2.89, 95% CI: 1.16–7.21; PRR = 2.88, x2 = 5.66), hypersomnia (ROR = 3.66, 95% CI: 1.45–9.22; PRR = 3.65, x2 = 8.67), and feeding disorder (ROR = 3.13, 95% CI: 1.12–8.75; PRR = 3.13, x2 = 5.29). These potential signals warrant attention in clinical practice.
Table 3.
Analysis of signal strength for treatment-emergent adverse events of FcRn antagonists at the Preferred Term (PT) level in the FAERS database.
| efgartigimod alfa |
Rozanolixizumab |
|||||||
|---|---|---|---|---|---|---|---|---|
| No | PT | n | ROR(95%Cl) | PRR(x2) | PT | n | ROR(95%Cl) | PRR(x2) |
| 1 | FALL | 173 | 2.03(1.69–2.43) | 2.01(61.62) | HEADACHE | 86 | 2.47(1.97–3.08) | 2.4(66.89) |
| 2 | URINARY TRACT INFECTION | 160 | 2.43(2–2.94) | 2.41(86.5) | DIARRHOEA | 47 | 2.31(1.71–3.12) | 2.28(31.96) |
| 3 | SYMPTOM RECURRENCE | 157 | 3.43(2.79–4.23) | 3.4(152.82) | VOMITING | 31 | 3.11(2.15–4.51) | 3.08(39.96) |
| 4 | CHOKING | 69 | 4.02(2.91–5.56) | 4.01(82.74) | HERPES ZOSTER | 11 | 2.32(1.26–4.29) | 2.32(7.71) |
| 5 | UPPER RESPIRATORY TRACT INFECTION | 55 | 2.78(1.98–3.88) | 2.77(38.61) | INJECTION SITE ERYTHEMA | 9 | 7.42(3.6–15.32) | 7.39(40.64) |
| 6 | NEPHROLITHIASIS | 48 | 3.57(2.45–5.22) | 3.56(49.6) | MUSCULOSKELETAL STIFFNESS | 9 | 2(1.02–3.93) | 2(4.24) |
| 7 | ATRIAL FIBRILLATION | 44 | 2.15(1.5–3.07) | 2.14(18.23) | SYNCOPE | 7 | 3.66(1.67–8) | 3.65(12.14) |
| 8 | DIVERTICULITIS | 36 | 2.87(1.89–4.35) | 2.86(26.72) | ADVERSE DRUG REACTION | 7 | 4.52(2.05–9.98) | 4.51(16.83) |
| 9 | DEHYDRATION | 33 | 2.23(1.47–3.39) | 2.23(15.02) | INJECTION SITE SWELLING | 6 | 5.65(2.37–13.44) | 5.63(19.53) |
| 10 | PRODUCTIVE COUGH | 26 | 2.31(1.44–3.71) | 2.31(12.78) | ADVERSE EVENT | 5 | 3.36(1.34–8.44) | 3.35(7.5) |
| 11 | DYSPNEA AT REST | 26 | 58.99(14–248.56) | 58.86(105.68) | JOINT SWELLING | 5 | 2.89(1.16–7.21) | 2.88(5.66) |
| 12 | RESPIRATORY TRACT INFECTION | 22 | 2.17(1.3–3.6) | 2.17(9.34) | HYPERSOMNIA | 5 | 2.74(1.1–6.84) | 2.74(5.1) |
| 13 | NEUROPATHY PERIPHERAL | 21 | 2.21(1.31–3.73) | 2.21(9.37) | DYSPEPSIA | 5 | 3.83(1.51–9.67) | 3.82(9.33) |
| 14 | RESPIRATORY SYNCYTIAL VIRUS INFECTION | 20 | 5.33(2.79–10.19) | 5.33(32.31) | MENINGITIS | 5 | 3.66(1.45–9.22) | 3.65(8.67) |
| 15 | LACRIMATION INCREASED | 18 | 2.63(1.47–4.71) | 2.63(11.5) | INJECTION SITE BRUISING | 5 | 8.67(3.23–23.23) | 8.65(26.78) |
| 16 | INJECTION SITE ERYTHEMA | 17 | 2.41(1.34–4.34) | 2.41(9.12) | EATING DISORDER | 4 | 3.13(1.12–8.75) | 3.13(5.29) |
| 17 | BLEPHAROSPASM | 16 | 2.59(1.4–4.79) | 2.59(9.92) | DISCOMFORT | 4 | 3.06(1.1–8.53) | 3.06(5.07) |
| 18 | STAPHYLOCOCCAL INFECTION | 16 | 2.07(1.15–3.74) | 2.07(6.08) | FLUID RETENTION | 4 | 2.8(1.01–7.78) | 2.8(4.26) |
| 19 | HEAD INJURY | 15 | 2.12(1.15–3.92) | 2.12(6.07) | INJECTION SITE PRURITUS | 4 | 5.06(1.77–14.52) | 5.05(11.28) |
| 20 | CARDIAC FAILURE CONGESTIVE | 15 | 2.19(1.18–4.06) | 2.19(6.55) | LOCALISED INFECTION | 4 | 3.37(1.2–9.45) | 3.37(6.05) |
| 21 | EYE PAIN | 14 | 2.35(1.23–4.48) | 2.35(7.14) | INFUSION SITE PRURITUS | 4 | 9.4(3.09–28.6) | 9.39(23.32) |
| 22 | BULBAR PALSY | 13 | 2.68(1.35–5.32) | 2.68(8.58) | TASTE DISORDER | 3 | 6.17(1.8–21.19) | 6.16(10.92) |
| 23 | PROSTATE CANCER | 13 | 2.56(1.3–5.06) | 2.56(7.89) | INJECTION SITE RASH | 3 | 4.29(1.29–14.3) | 4.29(6.69) |
| 24 | INJECTION SITE RASH | 13 | 4.53(2.1–9.78) | 4.53(17.87) | ||||
| 25 | INFUSION SITE EXTRAVASATION | 12 | 2.09(1.05–4.14) | 2.09(4.67) | ||||
| 26 | DYSURIA | 11 | 2.08(1.02–4.24) | 2.08(4.2) | ||||
| 27 | INJECTION SITE REACTION | 11 | 4.53(1.96–10.45) | 4.53(15.12) | ||||
| 28 | JOINT INJURY | 10 | 4.12(1.75–9.7) | 4.12(12.36) | ||||
| 29 | LUNG NEOPLASM MALIGNANT | 10 | 4.12(1.75–9.7) | 4.12(12.36) | ||||
| 30 | CLOSTRIDIUM DIFFICILE INFECTION | 10 | 2.52(1.16–5.45) | 2.52(5.87) | ||||
Subgroup analysis
This study investigated potential gender-based differences in adverse reactions between Efgartigimod alfa and Rozanolixizumab through gender subgroup analyses. According to Supplementary Tables S3 and S4, the most common adverse reactions in both male and female patients treated with Efgartigimod alfa were death, urinary tract infections, and infectious pneumonia. Notably, dyspnea exhibited the highest incidence in males (n = 27; ROR = 2.14, 95% CI: 1.44–3.19; PRR = 2.1, x2 = 14.73), whereas peripheral swelling was more frequently observed in female patients (ROR = 2.85, 95% CI: 1.23–6.58; PRR = 2.83, x2 = 6.56). Supplementary Tables S5 and S6 indicated that vomiting was the most common adverse reaction in both genders treated with Rozanolixizumab. Importantly, adverse reactions such as headache, meningitis, and heart failure were reported exclusively in males, while unique adverse events including herpes zoster, seizures, and hypersomnia occurred only in female patients. These findings suggest that gender-specific differences in adverse reactions should be carefully considered during clinical application of Efgartigimod alfa and Rozanolixizumab, with targeted monitoring and preventive measures implemented accordingly.
Time-to-onset analysis
As shown in Figure 1, most adverse events (AEs) associated with FcRn antagonists occurred within 30 days after treatment initiation. However, it is noteworthy that efgartigimod alfa continued to demonstrate a higher incidence of adverse events even after 180 days of treatment (>180 days). Furthermore, Weibull distribution analysis further validated the early failure pattern of this drug class, with detailed parameters provided in Table 4.
Figure 1.

Time to onset of adverse events in patients treated with FcRn antagonists. Most events occurred within the first 30 days after treatment initiation. Notably, a substantial number of reports for efgartigimod alfa were received beyond 180 days, while no such long-term reports were observed for rozanolixizumab.
Table 4.
Time to onset of adverse events associated with FcRn antagonists, and Weibull distribution analysis.
| Drug | TTO(days) |
Weibull distribution |
|||
|---|---|---|---|---|---|
| Case reports | Median(d)(IQR) | Scale parameter: α(95%CI) | Shape parameter: β(95%CI) | Type | |
| Efgartigimod alfa | 2,519 | 100(25.5–239) | 150.863(130.481–164.351) | 0.825(0.782–0.871) | Early failure |
| Rozanolixizumab | 442 | 18(3–60) | 37.256(26.388–52.599) | 0.617(0.529–0.720) | Early failure |
Discussion
In this retrospective pharmacovigilance analysis, we interrogated the FAERS database to characterize the postmarketing safety of the FcRn antagonists efgartigimod alfa and rozanolixizumab in real‑world clinical practice.20 This exploratory analysis aimed to identify potential safety signals using disproportionality methods. We extracted nearly two thousand adverse event reports and found that general disorders, nervous‑system symptoms and infections dominated the safety profiles. At the Preferred Term level, falls, urinary‑tract infections and symptom recurrence were leading complaints for efgartigimod alfa, whereas headache, diarrhea and vomiting predominated with rozanolixizumab. Importantly, our disproportionality analyses identified unexpected signals, including atrial fibrillation, peripheral neuropathy and prostate cancer for efgartigimod alfa, and meningitis, hypersomnia and feeding disorder for rozanolixizumab, that were not evident in pivotal trials. Most adverse events occurred within 30 days of treatment initiation, yet efgartigimod alfa displayed a tail of persistent risk beyond 180 days. We also uncovered distinct sex and age patterns: older adults and women accounted for the majority of efgartigimod reports, while rozanolixizumab reports clustered in adults aged 18–65 years; male patients taking rozanolixizumab experienced disproportionate rates of headache and meningitis, whereas female patients reported herpes zoster, seizures and hypersomnia. Collectively, these findings indicate that FcRn antagonists, though generally well tolerated, harbor specific safety signals and demographic susceptibilities that warrant personalized monitoring.21
When interpreting the results and comparing the two FcRn antagonists, it is crucial to consider the substantial disparity in the number of post-marketing reports (efgartigimod alfa: n = 2,519; rozanolixizumab: n = 442). This difference, likely influenced by factors such as earlier market entry and broader clinical experience with efgartigimod alfa, means that the safety profile of efgartigimod is more fully delineated in the current dataset. Consequently, the signals for rozanolixizumab should be interpreted with greater caution due to the smaller sample size and potentially lower statistical robustness. Our comparisons are intended to describe the emerging safety data for each agent individually rather than to directly rank their safety.
Because FcRn antagonists represent a novel, targeted immunotherapy, it is critical to contextualize their postmarketing safety within the broader landscape of myasthenia gravis treatment.22,23 FcRn mediates the salvage recycling of IgG; antagonizing the FcRn–IgG interaction accelerates IgG catabolism and lowers the concentration of pathogenic autoantibodies.24,25 FcRn inhibition therefore offers a mechanistically selective alternative to broad immunosuppression and plasma exchange.26 Our results align with this mechanism: infection‑related adverse events were among the most frequent complaints for both drugs, consistent with the theoretical risk of increased susceptibility to bacterial and viral pathogens.27 In the phase III ADAPT trial evaluating intravenous efgartigimod in patients with AChR-positive generalized myasthenia gravis, the most common treatment-emergent adverse events were headache, upper respiratory tract infections, and urinary tract infections.28 Similarly, in our FAERS analysis, urinary‑tract infection emerged as a leading preferred term for efgartigimod alfa and we detected elevated reporting odds ratios for upper respiratory‑tract infection and respiratory syncytial virus infection. The convergence between the pivotal trial and postmarketing data underscores the robustness of infection risk as an inherent feature of FcRn blockade. However, we also found signals for staphylococcal infection, diverticulitis and sepsis that were not documented in clinical trials, indicating that real‑world patients – often older and using concomitant immunosuppression – may be more vulnerable to opportunistic infections.
Our analysis diverges from the ADAPT trial in identifying cardiovascular and neurological signals absent from pre‑approval studies. Although the trial reported a low incidence of serious adverse events, our FAERS data highlighted atrial fibrillation and peripheral neuropathy as unexpected signals. The BMJ Open pharmacovigilance study likewise detected significant disproportionality for atrial fibrillation and transient ischemic attack.27 Mechanistically, FcRn antagonism is not known to perturb cardiac conduction; however, rapid IgG reduction could unmask underlying arrhythmic substrates in elderly patients or exacerbate comorbid autoimmune conditions.6,29 Peripheral neuropathy is another intriguing signal; IgG replacement therapy is known to alleviate neuropathic symptoms,30 and its depletion may conversely predispose to neural injury. While causal links remain speculative, these signals justify enhanced cardiovascular and neurological monitoring, especially in high‑risk patients. It is important to note that the observed association with conditions like atrial fibrillation and prostate cancer could also be influenced by the older age profile of the patient population treated with efgartigimod, as indicated in our descriptive analysis. Similarly, the association with prostate cancer raises questions about long‑term immunosurveillance; although oncological risk was not increased in clinical trials, IgG plays a role in tumor defense, and chronic suppression may theoretically impair immune surveillance.31
In contrast to efgartigimod alfa, the safety profile of rozanolixizumab in our analysis largely mirrored observations from the phase II and phase III trials. Headache was the predominant treatment‑emergent adverse event across trial doses. In the MycarinG phase III study, headache occurred in 45% of patients receiving 7 mg kg–1 rozanolixizumab, 38% receiving 10 mg kg–1, and 9% in the placebo group; diarrhea, pyrexia and nausea were also frequent, and serious adverse events were rare with no deaths or opportunistic infections.10 Our FAERS data reinforced this pattern: headache, diarrhea, vomiting and nausea were the top preferred terms, and we did not observe a clear infection signal. For rozanolixizumab, we identified potential signals for meningitis and hypersomnia, which were not prominently featured in clinical trials. The clinical significance of these signals is currently uncertain. The meningitis finding could indicate a rare susceptibility to CNS infections under IgG depletion, a phenomenon that theoretically could apply to the drug class, though reporting artifacts or misclassification cannot be ruled out. Similarly, hypersomnia may represent a novel, biologically plausible reaction to immunomodulation or could be related to the underlying disease or other factors. These potential signals underscore the need for continued surveillance. Although these signals were rare, they warrant prospective monitoring in ongoing long‑term trials.
Our dataset also allowed an exploratory analysis of demographic patterns. Among efgartigimod alfa reports, roughly three‑fifths originated from women and two‑thirds from patients over 65 years of age. In contrast, most rozanolixizumab reports came from adults aged 18–65 years, consistent with its more recent approval and use in a working‑age population. Notably, male patients receiving efgartigimod alfa were more likely to report dyspnea, whereas female patients reported peripheral swelling and urinary‑tract infection; this pattern parallels a survey of the Myasthenia Gravis registry, which showed that women experience more and more intolerable adverse effects from prednisone, including weight gain, mood swings, fatigue and headache.32 Such sex‑specific vulnerability may reflect pharmacokinetic differences, hormonal influences or disease severity and underscores the importance of sex‑stratified safety analyses. For rozanolixizumab, male‑specific adverse events included headache and meningitis, whereas female‑specific events encompassed herpes zoster, seizures and hypersomnia; these patterns suggest that differential immune responses to FcRn blockade may exist between sexes and highlight the need for personalized counseling.33
The temporal pattern of adverse events provides further insight into risk dynamics. In our study, most adverse events occurred within the first month of treatment, but efgartigimod alfa displayed a tail of events beyond six months, indicating a sustained but attenuated risk with ongoing therapy. The BMJ Open pharmacovigilance analysis found a median time to onset of 57 days, with 37% of events occurring by 30 days and 64% by 100 days, an early failure‑type pattern.27 This early clustering likely reflects the pharmacodynamic onset of IgG reduction and heightened clinical surveillance during initial cycles; however, persistence of later events emphasizes the importance of monitoring beyond the first cycle, particularly as patients transition to maintenance dosing or the subcutaneous formulation (Vyvgart Hytrulo). Real‑world data also revealed a sharp increase in efgartigimod alfa reports after the introduction of the subcutaneous formulation.27 At‑home administration broadens access but may also lead to rapid uptake in populations with comorbidities, underscoring the need for patient education and early reporting.
While our findings expand understanding of FcRn‑antagonist safety, several limitations should be acknowledged. FAERS is a spontaneous reporting system subject to under‑reporting, reporting bias and duplicate submissions, which preclude incidence calculations and causal inference. The denominator of exposed patients is unknown; thus, disproportionality analyses identify signals rather than risk estimates. Most importantly, this study is an exploratory pharmacovigilance analysis. The detected signals represent statistical associations based on reporting rates and should not be interpreted as proof of a causal relationship between the drug and the adverse event. Important confounders such as disease severity, concomitant immunosuppressants, vaccination status and comorbidities are not systematically captured, yet they can profoundly influence infection risk and other outcomes. Sex‑specific analyses are limited by missing data on pregnancy status and body mass index. Time‑to‑onset values are approximate to the nearest day, hindering analysis of immediate infusion reactions. In addition, our analysis cannot differentiate between intravenous and subcutaneous efgartigimod alfa, yet these formulations may differ in pharmacokinetics and safety. Lastly, signals such as prostate cancer could be coincidental due to longer life expectancy in treated patients; large prospective studies are necessary before inferring carcinogenicity.
Despite these limitations, our work has several implications for future research and clinical practice. First, identification of infection, atrial fibrillation and neuropathy signals emphasizes the need for baseline risk assessment and monitoring. Physicians should obtain histories of cardiac arrhythmias and neuropathies, ensure vaccinations and counsel patients regarding early signs of infection before initiating FcRn antagonists. Second, sex‑ and age‑specific patterns highlight the importance of tailored monitoring strategies. Prospective pharmacovigilance registries should stratify outcomes by sex, age and co‑medication to elucidate underlying mechanisms and refine dosing. Third, mechanistic studies are warranted to explore how IgG depletion may precipitate arrhythmias or neuropathies and whether concomitant immunosuppression modulates these risks. Preclinical models could investigate whether FcRn blockade alters autonomic function or cardiac conduction, while immunologic profiling may reveal biomarkers predicting susceptibility. Fourth, long‑term observational studies should evaluate the potential association between FcRn antagonists and malignancy or chronic comorbidities, given the theoretical concern of impaired tumor immunosurveillance. Fifth, as other FcRn‑targeted agents such as nipocalimab and batoclimab progress through clinical trials, comparative safety analyses will be essential to determine whether observed signals are class effects or molecule‑specific.34 Network meta‑analyses have already suggested that rozanolixizumab is associated with a higher incidence of headache than efgartigimod alfa and that cost per improved outcome may differ between agents;35 integrating real‑world safety data with efficacy and cost will inform treatment selection and reimbursement.
The emergence of FcRn antagonists underscores the importance of vigilant postmarketing surveillance for all novel immunotherapies. Traditional clinical trials are powered to detect common adverse events but often exclude older adults and patients with significant comorbidities. Real‑world pharmacovigilance complements trial data by capturing rare or demographically specific events and can inform labeling, risk management plans and clinical guidelines. As digital health tools expand, integrating electronic health records, patient‑reported outcomes and machine‑learning algorithms may enhance signal detection and provide more timely safety updates. In this evolving therapeutic landscape, collaboration among clinicians, regulators, patient advocacy groups and industry will be essential to ensure that FcRn antagonists realize their potential in transforming myasthenia gravis care while maintaining patient safety.
Conclusion
This study utilized the FAERS database to evaluate the safety of FcRn antagonists in the treatment of myasthenia gravis, employing two algorithms (ROR and PRR) for data analysis. In addition to confirming adverse reactions listed in the drug label, previously unreported potential adverse events were identified for each drug (e.g., atrial fibrillation and peripheral neuropathy for efgartigimod alfa; meningitis and hypersomnia for rozanolixizumab). However, as an exploratory disproportionality analysis, these findings indicate potential associations that warrant further investigation but do not prove causality. These findings should be interpreted with caution due to the inherent limitations of the FAERS database. Future validation through rigorous prospective clinical trials or epidemiological studies is recommended.
Supplementary Material
Acknowledgments
LBY and ZWC are co-first authors. Conceptualization: LBY, CDN. Data curation: LBY. Formal analysis: LBY, ZWC, HA. Funding acquisition: CDN. Investigation: LBY. Methodology: LBY. Software: LBY. Validation: LBY, CDN. Visualization: LBY. Writing - original draft: LBY. Writing - review & editing: LBY, CDN. All the authors read and approved the final version.
Biography
Danna Chen is an Associate Professor and Vice Dean of the School of Basic Medicine at Changsha Medical University. She has been recognized as a talent in the Hunan Provincial 121 Talent Program, an Outstanding Innovative Youth of Changsha, and a candidate for the Young Backbone Teacher Training Program in Hunan higher education institutions. She also serves as a council member of the Hunan Genetics Society. Her primary research focuses on genetics and reproduction.
Professor Chen has presided over more than 10 research projects, including grants from the National Natural Science Foundation of China, the Hunan Provincial Natural Science Foundation, and key projects of the Hunan Provincial Department of Education. She has also participated as a key researcher in several national-level initiatives, such as the National 973 Program and four projects supported by the National Natural Science Foundation of China.
With over 40 papers published in domestic and international academic journals (including more than 20 SCI-indexed articles), she has also co-authored a leading textbook published by People’s Medical Publishing House. Additionally, she was awarded the First Prize for Outstanding Achievements in Education and Teaching Reform by Hunan Province.
Funding Statement
The authors declare that the research, writing, and publication of this article received funding. This study was jointly supported by the following grants: Hunan Provincial Department of Education Fund Project [23A0667]; Changsha Outstanding Youth Innovation Talent Development Program [kq2106074]; National Undergraduate Innovation and Entrepreneurship Training Program 2024 (Letter No. 13 [2024] from the Department of Higher Education - [S202410823011]).
Disclosure statement
No potential conflict of interest was reported by the author(s).
Date availability statement
All original data can be accessed in The FDA Adverse Event Reporting System (FAERS) database (https://open.fda.gov/data/downloads/).
Supplementary material
Supplemental data for this article can be accessed online at https://doi.org/10.1080/21645515.2025.2586335
References
- 1.Dodd KC, Clay FJ, Forbes AM, Handley J, Keh RYS, Miller JA, Storms K, White LM, Lilleker JB, Sussman J.. Rituximab for myasthenia gravis. Cochrane Database Syst Rev. 2025;7(7):Cd014574. doi: 10.1002/14651858.CD014574.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Beeching F, Lecchi A, Riccitelli GC. Female gender and quality of life outcomes in myasthenia gravis: a systematic review and meta-analysis. Ther Adv Neurol Disord. 2025;18:17562864251344742. doi: 10.1177/17562864251344742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Giannopapas V, Stefanou MI, Zouvelou V, Papagiannopoulou G, Smyrni V, Kosmidou M, Kitsos DK, Keramida A, Salakou S, Tsivgoulis G, et al. Risk of diabetes mellitus in the myasthenia gravis: a systematic review and meta-analysis. J Clin Med. 2025;14(12):4221. doi: 10.3390/jcm14124221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.White LM, Clay FJ, Forbes AM, Keh RYS, Lilleker JB, Spillane J, Storms K, Dodd KC, Sussman J. FcRn inhibitors for myasthenia gravis. Cochrane Database Syst Rev. 2025;8(8):Cd016097. doi: 10.1002/14651858.CD016097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ma C, Shen J, Zhu Y, Zhu R. A prospective clinical trial of efgartigimod for new-onset generalized myasthenia gravis. Curr Neuropharmacol. 2025;23. doi: 10.2174/011570159X366227250716054651. [DOI] [PubMed] [Google Scholar]
- 6.Tarasco MC, Rinaldi E, Frangiamore R, Vanoli F, Berni A, Iacomino N, Canciello A, Andreetta F, Ciusani E, Bonanno S, et al. Unknown immunoregulatory effects of FcRn inhibition by efgartigimod in myasthenia gravis: a new mechanism of action beyond IgG reduction. Neurol Neuroimmunol Neuroinflamm. 2025;12(5):e200455. doi: 10.1212/NXI.0000000000200455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wu J, Zhu LN, Liu XY. Targeting neonatal Fc receptor blockade: reshaping the treatment landscape for IgG-mediated autoimmune diseases. Zhonghua Yi Xue Za Zhi. 2025;105(29):2440–12. doi: 10.3760/cma.j.cn112137-20250308-00560. [DOI] [PubMed] [Google Scholar]
- 8.Wu G, Xiang Y, Liu H, Hu C, Li Y, Feng J, Li Y. Virtual screening combined with phase transition-FRET for discovery of small-molecule FcRn antagonists. Anal Chem. 2025;97(20):10661–10670. doi: 10.1021/acs.analchem.5c00486. [DOI] [PubMed] [Google Scholar]
- 9.Heo YA. Efgartigimod alfa in generalised myasthenia gravis: a profile of its use. CNS Drugs. 2023;37(5):467–473. doi: 10.1007/s40263-023-01000-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Matic A, Bril V. Rozanolixizumab for myasthenia gravis: a breakthrough treatment and future prospects. Immunotherapy. 2025;17(5):309–316. doi: 10.1080/1750743X.2025.2491295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kiessling P, Lledo-Garcia R, Watanabe S, Langdon G, Tran D, Bari M, Christodoulou L, Jones E, Price G, Smith B, et al. The FcRn inhibitor rozanolixizumab reduces human serum IgG concentration: a randomized phase 1 study. Sci Transl Med. 2017;9(414). doi: 10.1126/scitranslmed.aan1208. [DOI] [PubMed] [Google Scholar]
- 12.Ulrichts P, Guglietta A, Dreier T, van Bragt T, Hanssens V, Hofman E, Vankerckhoven B, Verheesen P, Ongenae N, Lykhopiy V, et al. Neonatal Fc receptor antagonist efgartigimod safely and sustainably reduces IgGs in humans. J Clin Invest. 2018;128(10):4372–4386. doi: 10.1172/JCI97911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sakaeda T, Tamon A, Kadoyama K, Okuno Y. Data mining of the public version of the FDA adverse event reporting system. Int J Med Sci. 2013;10(7):796–803. doi: 10.7150/ijms.6048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Howard JF, Bril V, Vu T, Karam C, Peric S, Margania T, Murai H, Bilinska M, Shakarishvili R, Smilowski M, et al. Safety, efficacy, and tolerability of efgartigimod in patients with generalised myasthenia gravis (ADAPT): a multicentre, randomised, placebo-controlled, phase 3 trial. Lancet Neurol. 2021;20(7):526–536. doi: 10.1016/S1474-4422(21)00159-9. [DOI] [PubMed] [Google Scholar]
- 15.Saccà F, Barnett C, Vu T, Peric S, Phillips GA, Zhao S, Qi CZ, Gelinas D, Chiroli S, Verschuuren JJGM. Efgartigimod improved health-related quality of life in generalized myasthenia gravis: results from a randomized, double-blind, placebo-controlled, phase 3 study (ADAPT). J Neurol. 2023;270(4):2096–2105. doi: 10.1007/s00415-022-11517-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bril V, Drużdż A, Grosskreutz J, Habib AA, Kaminski HJ, Mantegazza R, Sacconi S, Utsugisawa K, Vu T, Boehnlein M, et al. Safety and efficacy of chronic weekly rozanolixizumab in generalized myasthenia gravis: the randomized open-label extension MG0004 study. J Neurol. 2025;272(4):275. doi: 10.1007/s00415-025-12958-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Brown EG. Using MedDRA: implications for risk management. Drug Saf. 2004;27(8):591–602. doi: 10.2165/00002018-200427080-00010. [DOI] [PubMed] [Google Scholar]
- 18.Rothman KJ, Lanes S, Sacks ST. The reporting odds ratio and its advantages over the proportional reporting ratio. Pharmacoepidemiol Drug Saf. 2004;13(8):519–523. doi: 10.1002/pds.1001. [DOI] [PubMed] [Google Scholar]
- 19.Sakaeda T, Kadoyama K, Minami K, Okuno Y. Commonality of drug-associated adverse events detected by 4 commonly used data mining algorithms. Int J Med Sci. 2014;11(5):461–465. doi: 10.7150/ijms.7967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lin J, Cheng Y, Chen L, Chen M, Shen Z. A real-world analysis of adverse event signals of cognitive and communication disorder in patients treated with Janus kinase inhibitors based on the FAERS database. J Am Acad Dermatol. 2025. doi: 10.1016/j.jaad.2025.08.003. [DOI] [PubMed] [Google Scholar]
- 21.Yang Y, Liu J, Wei W. A real-world pharmacovigilance study of efgartigimod alfa in the FDA adverse event reporting system database. Front Pharmacol. 2025;16:1510992. doi: 10.3389/fphar.2025.1510992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sivadasan A, Bril V. Clinical efficacy and safety of efgartigimod for treatment of myasthenia gravis. Immunotherapy. 2023;15(8):553–563. doi: 10.2217/imt-2022-0298. [DOI] [PubMed] [Google Scholar]
- 23.Gable KL, Guptill JT. Antagonism of the neonatal Fc receptor as an emerging treatment for myasthenia gravis. Front Immunol. 2019;10:3052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zhu LN, Hou HM, Wang S, Zhang S, Wang GG, Guo ZY, Wu J. FcRn inhibitors: a novel option for the treatment of myasthenia gravis. Neural Regen Res. 2023;18(8):1637–1644. doi: 10.4103/1673-5374.363824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ma G, Crowley AR, Heyndrickx L, Rogiers I, Parthoens E, Van Santbergen J, Ober RJ, Bobkov V, de Haard H, Ulrichts P, et al. Differential effects of FcRn antagonists on the subcellular trafficking of FcRn and albumin. JCI Insight. 2024;9(10). doi: 10.1172/jci.insight.176166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mina-Osorio P, Tran MH, Habib AA. Therapeutic plasma exchange versus FcRn inhibition in autoimmune disease. Transfus Med Rev. 2024;38(1):150767. doi: 10.1016/j.tmrv.2023.150767. [DOI] [PubMed] [Google Scholar]
- 27.Ju Y, Qin X. Postmarketing adverse events of efgartigimod alfa: a real-world pharmacovigilance study based on the FAERS database. BMJ Open. 2025;15(7):e095652. doi: 10.1136/bmjopen-2024-095652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Howard JF, Bril V, Vu T, Karam C, Peric S, De Bleecker JL, Murai H, Meisel A, Beydoun SR, Pasnoor M, et al. Long-term safety, tolerability, and efficacy of efgartigimod (ADAPT+): interim results from a phase 3 open-label extension study in participants with generalized myasthenia gravis. Front Neurol. 2023;14:1284444. doi: 10.3389/fneur.2023.1284444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mirza M, Strunets A, Shen WK, Jahangir A. Mechanisms of arrhythmias and conduction disorders in older adults. Clin Geriatr Med. 2012;28(4):555–573. doi: 10.1016/j.cger.2012.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Berger M, Allen JA. Optimizing IgG therapy in chronic autoimmune neuropathies: a hypothesis driven approach. Muscle Nerve. 2015;51(3):315–326. doi: 10.1002/mus.24526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Peter HH, Ochs HD, Cunningham-Rundles C, Vinh DC, Kiessling P, Greve B, Jolles S. Targeting FcRn for immunomodulation: benefits, risks, and practical considerations. J Allergy Clin Immunol. 2020;146(3):479–491.e5. doi: 10.1016/j.jaci.2020.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lee I, Kaminski HJ, McPherson T, Feese M, Cutter G. Gender differences in prednisone adverse effects: Survey result from the MG registry. Neurol Neuroimmunol Neuroinflamm. 2018;5(6):e507. doi: 10.1212/NXI.0000000000000507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Fiore NT, Willcox KF, Dayani D, Zuberi YA, Heijnen CJ, Grace PM. Reducing IgG accumulation via neonatal Fc receptor (FcRn) blockade relieves neuropathic pain. Brain Behav Immun. 2025;125:371–387. doi: 10.1016/j.bbi.2025.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Li J, Wu X, Chu T, Tan X, Wang S, Qu R, Chen Z, Wang Z. The efficacy and safety of FcRn inhibitors in patients with myasthenia gravis: a systematic review and meta-analysis. J Neurol. 2024;271(5):2298–2308. doi: 10.1007/s00415-024-12247-x. [DOI] [PubMed] [Google Scholar]
- 35.Cheng JJ, Wang FQ, Dai ZY, Wang XW, Wang Y. The efficacy and safety of efgartigimod for refractory myasthenia gravis: a systematic review and meta-analysis. Eur J Med Res. 2025;30(1):775. doi: 10.1186/s40001-025-03057-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
